US6737537B2 - Platinum (II) antitumor compounds - Google Patents

Platinum (II) antitumor compounds Download PDF

Info

Publication number
US6737537B2
US6737537B2 US10/146,971 US14697102A US6737537B2 US 6737537 B2 US6737537 B2 US 6737537B2 US 14697102 A US14697102 A US 14697102A US 6737537 B2 US6737537 B2 US 6737537B2
Authority
US
United States
Prior art keywords
hydroxy
alkyl
compound
cells
halo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US10/146,971
Other versions
US20030176410A1 (en
Inventor
Faith M. Uckun
Rama K. Narla
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hayashibara Seibutsu Kagaku Kenkyujo KK
Parker Hughes Institute
Original Assignee
Parker Hughes Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Parker Hughes Institute filed Critical Parker Hughes Institute
Priority to US10/146,971 priority Critical patent/US6737537B2/en
Assigned to KABUSHIKI KAISHA HAYASHIBARA SEIBUTSU KAGAKU KENKYUJO reassignment KABUSHIKI KAISHA HAYASHIBARA SEIBUTSU KAGAKU KENKYUJO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KINUGASA, KAZUSHI, MANDAI, SHINYA
Assigned to PARKER HUGHES INSTITUTE reassignment PARKER HUGHES INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NARLA, RAMA K., UCKUN, FATIH M.
Publication of US20030176410A1 publication Critical patent/US20030176410A1/en
Application granted granted Critical
Publication of US6737537B2 publication Critical patent/US6737537B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System compounds of the platinum group
    • C07F15/0086Platinum compounds
    • C07F15/0093Platinum compounds without a metal-carbon linkage

Definitions

  • the present invention relates to platinum (II) compounds effective for treating tumor cells and particularly effective to induce apoptosis in leukemia cells, breast cancer cells, prostate cancer cells, and brain cancer cells.
  • Cancer is a major disease that continues as one of the leading causes of death at any age. In the United States alone, it is anticipated that more than a half a million Americans will die of cancer in 2000. Currently, radiotherapy and chemotherapy are two important methods used in the treatment of cancer.
  • 4,661,516 discloses cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H 2 O
  • U.S. Pat. No. 4,758,588 discloses cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H 2 O
  • U.S. Pat. No. 5,011,959 discloses 1,1,-cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H 2 O
  • U.S. Pat. No. 5,132,323 discloses 1,1,-cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H 2 O.
  • the present invention describes new platinum (II) compounds which are particularly effective for treating cancer cells.
  • this invention in one aspect, relates to platinum (II) compounds including one or more analogues of formula (I):
  • R 1 and R 2 are independently halo
  • R 3 , R 4 , R 5 and R 6 are independently selected from the group consisting of: hydrogen, hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, and
  • R 7 and R 8 are independently selected from the group consisting of: hydroxy, (C 3 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 3 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl; or
  • R 7 and R 8 are independently selected from the group consisting of: hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • the invention relates to platinum (II) compounds of formula (I) above, wherein:
  • n 1 to 6
  • R 12 and R 13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, and
  • R 7 and R 8 are independently selected from the group consisting of: hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or m is 6 and R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl; or
  • R 12 and R 13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, and
  • R 7 and R 8 are independently selected from the group consisting of: hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • the present invention also relates to pharmaceutical compositions containing the aforementioned inventive compound(s) in combination with a pharmaceutically acceptable carrier.
  • the invention further provides a method of inducing apoptosis in a population of cells, including contacting the population of cells with the aforementioned inventive compound and/or composition(s) comprised of formula (I), wherein:
  • R 1 and R 2 are halo or when taken together form a group of the formula:
  • R 12 and R 13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • R 3 , R 4 , R 5 and R 6 are independently selected from the group consisting of: hydrogen, hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy and (C 1 -C 6 ) hydroxyalkyl; and
  • R 7 and R 8 are independently selected from the group consisting of: hydrogen, hydroxy, (C 1 -C 6 ) alkyl (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl, (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • the invention provides a method of treating a subject with a tumor, wherein the method comprises administering to the subject a therapeutically effective amount of the aforementioned compound(s) or composition(s).
  • FIG. 1 shows experimental results for Example 3, including the cell survival rates for differing drug concentrations of the inventive compounds for NALM-6 cells (FIG. 1 a ) and MOLT-3 cells (FIG. 1 b ).
  • FIG. 2 shows experimental results for Example 4, including the cell survival rates for differing drug concentrations of the inventive compounds incubated for 24 hours (FIG. 2 a ), 48 hours (FIG. 2 b ), 72 hours (FIG. 2 c ), and 96 hours (FIG. 2 d ).
  • halogen refers to bromine, chlorine, fluorine, and iodine.
  • alkyl refers to a branched or unbranched saturated hydrocarbon group, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, octyl, decyl, tetradecyl, hexadecyl, eicosyl, tetracosyl and the like.
  • the alkyl group may have one or more hydrogen atoms replaced with a functional group.
  • cycloalkane refers to a cyclic alkane group.
  • alkoxy intends an alkyl group bound through a single, terminal ether linkage; that is, an “alkoxy” group may be defined as —OR where R is alkyl as defined above.
  • a “lower alkoxy” group intends an alkoxy group containing from one to six, more preferably from one to four, carbon atoms.
  • alkenyl refers to a branched or unbranched mon-unsaturated or di-unsaturated hydrocarbon group, which may have one or more hydrogen atoms replaced with a functional group. Geometric structures such as (AB)C ⁇ C(CD) are intended to include all isomers.
  • cycloalkenyl refers to a cyclic alkenyl group.
  • aryl refers to a C 6 H 6 aromatic ring. Substituents on the aryl group may be present on any position, i.e. ortho, meta or para positions or fused to the aromatic ring.
  • hydroxyalkyl is meant a branched, unbranched, or cyclic saturated hydrocarbon group with a terminal hydroxy group, such as methanol, ethanol,n-propanol, isopropanol, n-butanol, isobutanol, t-butanol, octanol, decanol, tetradecanol, hexadecanol and the like.
  • the alkyl group may have one or more hydrogen atoms replaced with a functional group.
  • platinum (II) compound or “platinum (II) analogue” it is meant to include compounds of the formula (I):
  • R 1 and R 2 are halo or when together form a group of the formula:
  • R 12 and R 13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • R 3 , R 4 , R 5 and R 6 are independently selected from the group consisting of: hydrogen, hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl; and
  • R 7 and R 8 are independently selected from the group consisting of: hydrogen, hydroxy, (C 1 -C 6 ) alkyl, (C 3 -C 6 ) cycloalkyl, (C 2 -C 6 ) alkenyl, (C 3 -C 6 ) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl, or when taken together form a group of the formula:
  • R 10 and R 11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C 1 -C 6 ) alkyl (C 2 -C 6 ) alkenyl, (C 1 -C 6 ) alkoxy, and (C 1 -C 6 ) hydroxyalkyl;
  • apoptosis is meant cell death, inhibition of clonogenic growth, and cytotoxicity. Therefore, inducing apoptosis includes inducing cell death, inhibiting clonogenic growth, and inducing cytotoxicity.
  • contacting is meant an instance of exposure of at least one cell (e.g., a neural cell, a stem cell, a cardiac cell) to an agent (e.g., a platinum (II) compound).
  • a cell e.g., a neural cell, a stem cell, a cardiac cell
  • agent e.g., a platinum (II) compound
  • cancer or “tumor” includes cancers of the head and neck, lung, mesothelioma, mediastinum, esophagus, stomach, pancreas, hepatobiliary system, small intestine, colon, rectum, anus, kidney, ureter, bladder, prostate, urethra, penis, testis, gynecological organs, ovarian, breast, endocrine system, skin central nervous system; sarcomas of the soft tissue and bone; myeloma; melanoma of cutaneous and intraocular origin; childhood leukemia and lymphomas, Hodgkin's disease, lymphomas of lymphocytic and cutaneous origin, acute and chronic leukemia, plasma cell neoplasm and cancers associated with AIDS.
  • cancer is not Sarcoma 180.
  • the preferred mammalian species for treatment are humans and domesticated animals.
  • the term “subject” is meant an individual.
  • the subject is a mammal such as a primate, and more preferably, a human.
  • the “subject” can include domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), and laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.).
  • terapéuticaally effective amount or “therapeutically effective dose” means the amount needed to achieve the desired result or results (reducing or delaying apoptosis or treating a degenerative condition).
  • a “therapeutically effective amount” can vary for the various platinum (II) compounds used in the invention.
  • One skilled in the art can readily assess the potency of the compounds.
  • pharmaceutically acceptable is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the selected bicyclic compound without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ⁇ -ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • Representative pharmaceutically acceptable bases are ammonium hydroxide, sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, magnesium hydroxide, ferrous hydroxide, zinc hydroxide, copper hydroxide, aluminum hydroxide, ferric hydroxide, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, lysine, arginine, histidine, and the like.
  • the reaction is conducted in water, alone or in combination with an inert, water-miscible organic solvent, at a temperature of from about 0° C. to 100° C., preferably at room temperature.
  • the molar ratio of compounds of structural formula (I) to base used are chosen to provide the ratio desired for any particular salts.
  • the starting material can be treated with approximately one equivalent of base to yield a salt.
  • approximately one-half a molar equivalent of base is used to yield a neutral salt, while for aluminum slats, approximately one-third a molar equivalent of base will be used.
  • Ester derivatives are typically prepared as precursors to the acid form of the compounds, and accordingly may serve as prodrugs. Generally, these derivatives will be alkyl esters such as methyl, ethyl, and the like. Amide derivatives/—(CO)NH 2 , —(CO)NHR and —(CO)NR 2 , where R is alkyl, may be prepared by reaction of the carboxylic acid-containing compound with ammonia or a substituted amine.
  • n is preferably 1 to 3, more preferably 3;and R 12 and R 13 are hydrogen, halo, or hydroxy, more preferably hydrogen.
  • R 3 , R 4 , R 5 , and R 6 are hydrogen, hydroxy, and C 1 -C 6 alkyl, more preferably hydrogen or C 1 -C 3 alkyl.
  • R 7 and R 8 are hydrogen, C 1 -C 6 alkyl, or R 7 and R 8 when taken together form a group of the formula:
  • m is preferably 1 to 6 and R 10 and R 11 are preferably hydrogen, C 1 -C 6 alkyl or halo, most preferably hydrogen.
  • Particularly useful compounds of formula (I) include:
  • the compounds of the present invention may be readily synthesized using techniques generally known to synthetic organic chemists. Suitable experimental methods for making and derivatizing aromatic compounds are described, for example, in the references cited in the Background section herein, the disclosures of which are hereby incorporated by reference for their general teachings and for their synthesis teachings. Although any known synthetic route may be used, preferred routes are illustrated by way of example in scheme 1.
  • the compounds of formula (I) as above defined may be useful in inducing apoptosis in a population of cells, and/or treating a subject with a tumor.
  • the methods include contacting the cells with a compound or composition of formula (I) as defined above, or administering to the subject a therapeutically effective amount of a compound or composition of formula (I).
  • the cells are lymphoblastic leukemia cells, multiple myeloma cells, breast cancer cells, prostate cancer cells, brain tumor cells, neural tumor cells, or neuronal tumor cells.
  • the tumor cells include malignant and benign tumor cells.
  • the apoptosis would result in at least a 10% reduction in the number of cells, including, for example, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any amount in between.
  • the reduction can be measured, for example, by comparing the number of cells after contact with the platinum (II) compound to the number of cells in a jig control population of cells lacking contact with the platinum (II) compound. Histological signs of apoptosis in cells after contact with the platinum (II) compound include condensation of the chromatin, the occurrence of apoptotic bodies, and cellular shrinkage. DNA laddering and other signs of DNA degradation are also signs of apoptosis. Apoptosis can also be assessed indirectly by observing, for example, a reduction in the amount of release or activity by the population of cells. Thus, if the cell population undergoes apoptosis, a decrease in neurotransmitter release upon stimulation or neuronal cells would decrease.
  • the cells can be contacted in vitro with the platinum (II) compound, for example, by adding the compound to the culture medium (by continuous infusion, by bolus delivery, or by changing the medium to a medium that contains the agent) or by adding the agent to the extracellular fluid in vivo (by local delivery, systemic delivery, intravenous injection, bolus delivery, or continuous infusion).
  • the duration of “contact” with a cell or population of cells is determined by the time the compound is present at physiologically effective levels or at presumed physiologically effective levels in the medium or extracellular fluid bathing the cell or cells.
  • the duration of contact is 1-96 hours, and more preferably, for 24 hours, but such time would vary based on the half life of the compound and could be optimized by one skilled in the art using routine experimentation.
  • the platinum (II) compounds can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient or a domestic animal in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the platinum (II) compounds of the present invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety.
  • gene therapy methods of delivery See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety.
  • primary cells transfected with the gene for the compound of the present invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells.
  • the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • compositions and preparations may, of odurse, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form.
  • amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, dragees, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • the active compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compounds of formula I to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compounds of formula I can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the concentration of the compound(s) of formula I in a liquid composition will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%.
  • concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the compound varies depending on the target cell, tumor, tissue, graft, or organ.
  • a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
  • the compound may conveniently be administered in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
  • the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 to about 75 ⁇ M, preferably, about 1 to 50 ⁇ M, most preferably, about 2 to about 30 ⁇ M. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infuisions containing about 0.4-15 mg/kg of the active ingredient(s).
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • An administration regimen could include long-term, daily treatment.
  • long-term is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E. W., ed. 4), Mack Publishing Co., Easton, Pa. The dosage can also be adjusted by the individual physician in the event of any complication.
  • Compound of formula I a compound of formula I (‘Compound of formula I’)
  • the following illustrate representative pharmaceutical dosage forms, containing a compound of formula I (‘Compound of formula I’), for therapeutic or prophylactic use in humans.
  • Aerosol mg/can Compound of formula I 20.0 Oleic acid 10.0 Trichloromonofluoromethane 5,000.0 Dichlorodifluoromethane 10,000.0 Dichlorotetrafluoroethane 5,000.0
  • the effectiveness of inducing apoptosis by compound (5) as synthesized and described in example 2 was measured for human B-lineage acute lymphoblastic leukemia (NALM-6) cells, T-lineage acute lymphoblastic leukemia (MOLT-3) cells, and multiple myeloma (ARH-77) cells.
  • the cells were incubated with concentrations of 5 ⁇ M of compound (5) in 96-well plates 48 hours.
  • An in situ apoptosis assay which allows the detection of exposed 3′-hydroxyl groups in fragmented DNA by TdT-mediated dUTP nick-end labeling (TUNEL), was used by fixing, permeabilizing and visualizing the DNA degradation. Apoptotic nuclei were detected visually with a color change.
  • the amount of apoptotic nuclei of the control cells (not incubated with compound (5)) and the incubated cells were compared. In all cell lines, the compound (5) greatly increased apoptosis over the control.
  • the effect on clonogenic growth of human cancer cell lines was determined for compound (5) as synthesized and described in example 2 and cisplastin for human B-lineage acute lymphoblastic leukemia (NALM-6) cells and T-lineage acute lymphoblastic leukemia (MOLT-3) cells.
  • the cells were incubated with concentrations ranging from 0 (control) to 10 ⁇ M of the appropriate compound for 24 hours at 37° C. in a humidified 5% CO 2 atmosphere.
  • the cells were then resuspended in clonogenic a-MEM medium containing 0.9% methyl cellulose.
  • 1 ⁇ 10 4 cells/dish were plated in 35 mm Petri dishes and cultured for 7 days at 37° C. in a humidified 5% CO 2 atmosphere.
  • the colonies containing ⁇ 20 cells were counted using an inverted phase microscope. The results are shown below in table 4.

Abstract

The present invention describes platinum (II) compounds and compositions useful for treating a subject with a tumor and/or inducing apoptosis in a population of cells. The present invention also describes pharmaceutical compositions containing the aforementioned inventive compound(s) in combination with a pharmaceutically acceptable carrier. Additionally, the invention further provides a method of inducing apoptosis in a population of cells and a method of treating a subject with a tumor, wherein the method comprises administering to the subject a therapeutically effective amount of the aforementioned compound(s) or composition(s).

Description

This application is a continuation application of international application Ser. No. PCT/US00/31297 filed on Nov. 15, 2000 claiming priority under 35 U.S.C. 119 (a)-(e) to U.S. Provisional Application No. 60/165,652 filed on Nov. 15, 1999; the international application was published under PCT article 21(2) in English as WO 36431.
FIELD OF THE INVENTION
The present invention relates to platinum (II) compounds effective for treating tumor cells and particularly effective to induce apoptosis in leukemia cells, breast cancer cells, prostate cancer cells, and brain cancer cells.
BACKGROUND OF THE INVENTION
Cancer is a major disease that continues as one of the leading causes of death at any age. In the United States alone, it is anticipated that more than a half a million Americans will die of cancer in 2000. Currently, radiotherapy and chemotherapy are two important methods used in the treatment of cancer.
Considerable efforts are underway to develop new chemotherapeutic agents for more potent and specific anti-cancer therapy, presenting effective and efficient cytotoxicity against tumor cells, with minimal interference with normal cell function. Accordingly, there is an urgent need for the development and analysis of novel, effective anti-cancer agents.
The use of platinum (II) compounds as anti-cancer agents has been reported. For example, Cleare et al, Bioorganic Chem., 2, 187-210 (1973) discloses Pt2I2(EA)2 and Pt2Cl2(EA)2; Friedman et al discloses cis-Pt(NH2ETOH)2Cl2; Zimmerman et al, Inorganica Chimica Acta, 292 (1999) 127-130 discloses Pt2I2(EA)2 and Pt2Cl2(EA)2; U.S. Pat. No. 4,661,516 discloses cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H2O; U.S. Pat. No. 4,758,588 discloses cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H2O; U.S. Pat. No. 5,011,959 discloses 1,1,-cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H2O; and U.S. Pat. No. 5,132,323 discloses 1,1,-cyclopropanedicarboxylato(trans-R,R-DACH) Pt(II)H2O. The present invention describes new platinum (II) compounds which are particularly effective for treating cancer cells.
SUMMARY OF THE INVENTION
In accordance with the purpose(s) of this invention, as embodied and broadly described herein, this invention, in one aspect, relates to platinum (II) compounds including one or more analogues of formula (I):
Figure US06737537-20040518-C00001
wherein:
R1 and R2 are independently halo,
R3, R4, R5 and R6 are independently selected from the group consisting of: hydrogen, hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, and
a) when R1=R2
R7 and R8 are independently selected from the group consisting of: hydroxy, (C3-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C3-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00002
wherein m is 1 to 5 and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl; or
b) when R1≠R2,
R7 and R8 are independently selected from the group consisting of: hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00003
wherein m is 1 to 6 and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
or a pharmaceutically acceptable salt or ester thereof.
In a second aspect, the invention relates to platinum (II) compounds of formula (I) above, wherein:
R1 and R2 when taken together form a group of the formula:
Figure US06737537-20040518-C00004
wherein: n is 1 to 6, and
a) when n=1 to 2,
R12 and R13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, and
R7 and R8 are independently selected from the group consisting of: hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00005
wherein m is 1 to 5 and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or m is 6 and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl; or
b) when n=3 to 6,
R12 and R13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, and
R7 and R8 are independently selected from the group consisting of: hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00006
wherein m is 1 to 6 and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
or a pharmaceutically acceptable salt or ester thereof.
Moreover, the present invention also relates to pharmaceutical compositions containing the aforementioned inventive compound(s) in combination with a pharmaceutically acceptable carrier.
The invention further provides a method of inducing apoptosis in a population of cells, including contacting the population of cells with the aforementioned inventive compound and/or composition(s) comprised of formula (I), wherein:
R1 and R2 are halo or when taken together form a group of the formula:
Figure US06737537-20040518-C00007
wherein n is 1 to 6, and R12 and R13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
R3, R4, R5 and R6 are independently selected from the group consisting of: hydrogen, hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy and (C1-C6) hydroxyalkyl; and
R7 and R8 are independently selected from the group consisting of: hydrogen, hydroxy, (C1-C6) alkyl (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00008
wherein m is 1 to 6, and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
or a pharmaceutically acceptable salt or ester thereof.
In yet another aspect, the invention provides a method of treating a subject with a tumor, wherein the method comprises administering to the subject a therapeutically effective amount of the aforementioned compound(s) or composition(s).
Additional advantages of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention. The advantages of the invention will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate several experimental examples and together with the description, serve to explain the principles of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows experimental results for Example 3, including the cell survival rates for differing drug concentrations of the inventive compounds for NALM-6 cells (FIG. 1a) and MOLT-3 cells (FIG. 1b).
FIG. 2 shows experimental results for Example 4, including the cell survival rates for differing drug concentrations of the inventive compounds incubated for 24 hours (FIG. 2a), 48 hours (FIG. 2b), 72 hours (FIG. 2c), and 96 hours (FIG. 2d).
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention may be understood more readily by reference to the following detailed description of preferred embodiments of the invention and the Examples included therein and to the Figures and their previous and following description.
Before the present compounds, compositions, and/or methods are disclosed and described, it is to be understood that this invention is not limited to specific synthetic methods of using or making which may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
In this specification and in the claims that follow, reference will be made to a number of terms which shall be defined to have the following meanings:
Reference in the specification and concluding claims to parts by weight of a particular component in a composition, denotes the weight relationship between the component and any other components in the composition for which a part by weight is expressed.
The term “halogen” or “halo” refers to bromine, chlorine, fluorine, and iodine.
The term “alkyl” as used herein refers to a branched or unbranched saturated hydrocarbon group, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, octyl, decyl, tetradecyl, hexadecyl, eicosyl, tetracosyl and the like. The alkyl group may have one or more hydrogen atoms replaced with a functional group. The term “cycloalkane” as used herein refers to a cyclic alkane group.
The term “alkoxy” as used herein intends an alkyl group bound through a single, terminal ether linkage; that is, an “alkoxy” group may be defined as —OR where R is alkyl as defined above. A “lower alkoxy” group intends an alkoxy group containing from one to six, more preferably from one to four, carbon atoms.
The term “alkenyl” as used herein refers to a branched or unbranched mon-unsaturated or di-unsaturated hydrocarbon group, which may have one or more hydrogen atoms replaced with a functional group. Geometric structures such as (AB)C═C(CD) are intended to include all isomers. The term “cycloalkenyl” as used herein refers to a cyclic alkenyl group.
The term “aryl” as used herein refers to a C6H6 aromatic ring. Substituents on the aryl group may be present on any position, i.e. ortho, meta or para positions or fused to the aromatic ring.
By “hydroxyalkyl” is meant a branched, unbranched, or cyclic saturated hydrocarbon group with a terminal hydroxy group, such as methanol, ethanol,n-propanol, isopropanol, n-butanol, isobutanol, t-butanol, octanol, decanol, tetradecanol, hexadecanol and the like. The alkyl group may have one or more hydrogen atoms replaced with a functional group.
By “platinum (II) compound” or “platinum (II) analogue” it is meant to include compounds of the formula (I):
Figure US06737537-20040518-C00009
wherein:
R1 and R2 are halo or when together form a group of the formula:
Figure US06737537-20040518-C00010
wherein n is 1 to 6, and R12 and R13 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
R3, R4, R5 and R6 are independently selected from the group consisting of: hydrogen, hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl; and
R7 and R8 are independently selected from the group consisting of: hydrogen, hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, aryl which may be unsubstituted or substituted with a halo, hydroxy, or (C1-C6) alkyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl, or when taken together form a group of the formula:
Figure US06737537-20040518-C00011
wherein m is 1 to 6, and R10 and R11 are independently selected from the group consisting of: hydrogen, halo, hydroxy, (C1-C6) alkyl (C2-C6) alkenyl, (C1-C6) alkoxy, and (C1-C6) hydroxyalkyl;
or a pharmaceutically acceptable salt or ester thereof.
By “apoptosis” is meant cell death, inhibition of clonogenic growth, and cytotoxicity. Therefore, inducing apoptosis includes inducing cell death, inhibiting clonogenic growth, and inducing cytotoxicity.
As used throughout, by “contacting” is meant an instance of exposure of at least one cell (e.g., a neural cell, a stem cell, a cardiac cell) to an agent (e.g., a platinum (II) compound).
As used herein, the term “cancer” or “tumor” includes cancers of the head and neck, lung, mesothelioma, mediastinum, esophagus, stomach, pancreas, hepatobiliary system, small intestine, colon, rectum, anus, kidney, ureter, bladder, prostate, urethra, penis, testis, gynecological organs, ovarian, breast, endocrine system, skin central nervous system; sarcomas of the soft tissue and bone; myeloma; melanoma of cutaneous and intraocular origin; childhood leukemia and lymphomas, Hodgkin's disease, lymphomas of lymphocytic and cutaneous origin, acute and chronic leukemia, plasma cell neoplasm and cancers associated with AIDS. In one embodiment, cancer is not Sarcoma 180. The preferred mammalian species for treatment are humans and domesticated animals.
The term “subject” is meant an individual. Preferably, the subject is a mammal such as a primate, and more preferably, a human. Thus, the “subject” can include domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), and laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.).
In general, “therapeutically effective amount” or “therapeutically effective dose” means the amount needed to achieve the desired result or results (reducing or delaying apoptosis or treating a degenerative condition). One of ordinary skill in the art will recognize that the potency and, therefore, a “therapeutically effective amount” can vary for the various platinum (II) compounds used in the invention. One skilled in the art can readily assess the potency of the compounds.
By “pharmaceutically acceptable” is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the selected bicyclic compound without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Representative pharmaceutically acceptable bases are ammonium hydroxide, sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, magnesium hydroxide, ferrous hydroxide, zinc hydroxide, copper hydroxide, aluminum hydroxide, ferric hydroxide, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, lysine, arginine, histidine, and the like. The reaction is conducted in water, alone or in combination with an inert, water-miscible organic solvent, at a temperature of from about 0° C. to 100° C., preferably at room temperature. The molar ratio of compounds of structural formula (I) to base used are chosen to provide the ratio desired for any particular salts. For preparing, for example, the ammonium salts of the free acid starting material, a particular preferred embodiment, the starting material can be treated with approximately one equivalent of base to yield a salt. When calcium salts are prepared, approximately one-half a molar equivalent of base is used to yield a neutral salt, while for aluminum slats, approximately one-third a molar equivalent of base will be used.
Ester derivatives are typically prepared as precursors to the acid form of the compounds, and accordingly may serve as prodrugs. Generally, these derivatives will be alkyl esters such as methyl, ethyl, and the like. Amide derivatives/—(CO)NH2, —(CO)NHR and —(CO)NR2, where R is alkyl, may be prepared by reaction of the carboxylic acid-containing compound with ammonia or a substituted amine.
It will be appreciated by those skilled in the art that compounds of the invention having a chiral center may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein, it being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
Specific values listed below for radicals, substituents, and ranges, are for illustration only; they do not exclude other defined values or other values within defined ranges for the radicals and substituents.
Preferred constituents of R1 and R2 for the compounds of formula I are: where R1=R2, Cl, Br, or I, or where R1 and R2 when taken together form a group of the formula:
Figure US06737537-20040518-C00012
n is preferably 1 to 3, more preferably 3;and R12 and R13 are hydrogen, halo, or hydroxy, more preferably hydrogen.
Preferred constituents of R3, R4, R5, and R6 are hydrogen, hydroxy, and C1-C6 alkyl, more preferably hydrogen or C1-C3 alkyl.
Preferred constituents of R7 and R8 are hydrogen, C1-C6 alkyl, or R7 and R8 when taken together form a group of the formula:
Figure US06737537-20040518-C00013
wherein m is preferably 1 to 6 and R10 and R11 are preferably hydrogen, C1-C6 alkyl or halo, most preferably hydrogen.
Particularly useful compounds of formula (I) include:
Figure US06737537-20040518-C00014
Synthetic Methods
The compounds of the present invention may be readily synthesized using techniques generally known to synthetic organic chemists. Suitable experimental methods for making and derivatizing aromatic compounds are described, for example, in the references cited in the Background section herein, the disclosures of which are hereby incorporated by reference for their general teachings and for their synthesis teachings. Although any known synthetic route may be used, preferred routes are illustrated by way of example in scheme 1.
Figure US06737537-20040518-C00015
Utility and Administration
The compounds of formula (I) as above defined may be useful in inducing apoptosis in a population of cells, and/or treating a subject with a tumor.
The methods include contacting the cells with a compound or composition of formula (I) as defined above, or administering to the subject a therapeutically effective amount of a compound or composition of formula (I). In one embodiment, the cells are lymphoblastic leukemia cells, multiple myeloma cells, breast cancer cells, prostate cancer cells, brain tumor cells, neural tumor cells, or neuronal tumor cells. The tumor cells include malignant and benign tumor cells.
It is well known in the art how to determine antitumor activity or cytotoxicity using the standard tests described herein, or using other similar tests. Preferably, the apoptosis would result in at least a 10% reduction in the number of cells, including, for example, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any amount in between.
The reduction can be measured, for example, by comparing the number of cells after contact with the platinum (II) compound to the number of cells in a jig control population of cells lacking contact with the platinum (II) compound. Histological signs of apoptosis in cells after contact with the platinum (II) compound include condensation of the chromatin, the occurrence of apoptotic bodies, and cellular shrinkage. DNA laddering and other signs of DNA degradation are also signs of apoptosis. Apoptosis can also be assessed indirectly by observing, for example, a reduction in the amount of release or activity by the population of cells. Thus, if the cell population undergoes apoptosis, a decrease in neurotransmitter release upon stimulation or neuronal cells would decrease.
The cells can be contacted in vitro with the platinum (II) compound, for example, by adding the compound to the culture medium (by continuous infusion, by bolus delivery, or by changing the medium to a medium that contains the agent) or by adding the agent to the extracellular fluid in vivo (by local delivery, systemic delivery, intravenous injection, bolus delivery, or continuous infusion). The duration of “contact” with a cell or population of cells is determined by the time the compound is present at physiologically effective levels or at presumed physiologically effective levels in the medium or extracellular fluid bathing the cell or cells. Preferably, the duration of contact is 1-96 hours, and more preferably, for 24 hours, but such time would vary based on the half life of the compound and could be optimized by one skilled in the art using routine experimentation.
The platinum (II) compounds can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient or a domestic animal in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
The platinum (II) compounds of the present invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Pat. No. 5,399,346, which is incorporated by reference in its entirety. Using a gene therapy method of delivery, primary cells transfected with the gene for the compound of the present invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells.
Thus, the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of odurse, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
The tablets, dragees, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.
The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form must be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
For topical administration, the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
Examples of useful dermatological compositions which can be used to deliver the compounds of formula I to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
Useful dosages of the compounds of formula I can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
Generally, the concentration of the compound(s) of formula I in a liquid composition, such as a lotion, will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the compound varies depending on the target cell, tumor, tissue, graft, or organ.
In general, however, a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
The compound may conveniently be administered in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
Ideally, the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 to about 75 μM, preferably, about 1 to 50 μM, most preferably, about 2 to about 30 μM. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infuisions containing about 0.4-15 mg/kg of the active ingredient(s).
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
An administration regimen could include long-term, daily treatment. By “long-term” is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E. W., ed. 4), Mack Publishing Co., Easton, Pa. The dosage can also be adjusted by the individual physician in the event of any complication.
Experimental
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices, and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.) but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric.
EXAMPLE 1
The following illustrate representative pharmaceutical dosage forms, containing a compound of formula I (‘Compound of formula I’), for therapeutic or prophylactic use in humans.
(i) Tablet 1 mg/tablet
Compound of formula I 100.0
Lactose 77.5
Povidone 15.0
Croscarmellose sodium 12.0
Microcrystalline cellulose 92.5
Magnesium stearate 3.0
300.0
(ii) Tablet 2 mg/tablet
Compound of formula I 20.0
Microcrystalline cellulose 410.0
Starch 50.0
Sodium starch glycolate 15.0
Magnesium stearate 5.0
500.0
(iii) Capsule mg/capsule
Compound of formula I 10.0
Colloidal silicon dioxide 1.5
Lactose 465.5
Pregelatinized starch 120.0
Magnesium stearate 3.0
600.0
(iv) Injection 1 (1 mg/ml) mg/ml
Compound of formula I (free acid form) 1.0
Dibasic sodium phosphate 12.0 
Monobasic sodium phosphate 0.7
Sodium chloride 4.5
1.0 N Sodium hydroxide solution q.s.
(pH adjustment to 7.0-7.5)
Water for injection q.s. ad 1 mL
(v) Injection 2 (10 mg/ml) mg/ml
Compound of formula I (free acid form) 10.0
Monobasic sodium phosphate 0.3
Dibasic sodium phosphate 1.1
Polyethylene glycol 400 200.0
01 N Sodium hydroxide solution q.s.
(pH adjustment to 7.0-7.5)
Water for injection q.s. ad 1 mL
(vi) Aerosol mg/can
Compound of formula I 20.0
Oleic acid 10.0
Trichloromonofluoromethane 5,000.0
Dichlorodifluoromethane 10,000.0
Dichlorotetrafluoroethane 5,000.0
The above formulations may be obtained by conventional procedures well known in the pharmaceutical art.
EXAMPLE 2
The following compounds of formula I were synthesized via scheme 1.
(1) DDE246: cis-[PtI2(NH2C2H4OH)2] with the chemical formula (1):
Figure US06737537-20040518-C00016
(2) DDE247: cis-[PtCl2(NH2C2H4OH)2] with the chemical formula (2):
Figure US06737537-20040518-C00017
(3) DDE248: cis-[Pt(CPDCA)(NH2C2H4OH)2] with the chemical formula (3):
Figure US06737537-20040518-C00018
(4) DDE249: cis-[PtI2(NH2C3H6OH )2] with the chemical formula (4):
Figure US06737537-20040518-C00019
(5) DDE250: cis-[Pt(CPDCA)(DACH)]H2O with the chemical formula (5):
Figure US06737537-20040518-C00020
(6) DDE173: cis-[Pt(CPDCA)(NH3)2]H2O with the chemical formula (6):
Figure US06737537-20040518-C00021
Wherein
CPDCA=cyclopropane-1,1-dicarboxylic acid
DACH=trans-1,2-diaminocyclohexane
Elemental analysis was performed on the compounds resulting in the values shown in table 1.
TABLE 1
Compound C found (calc) H found (calc) N found (calc)
(1) DDE246 8.53 (8.41) 2.45 (2.47) 4.91 (4.91)
(2) DDE247 12.37 (12.38) 3.62 (3.64) 7.05 (7.22)
(3) DDE248 24.46 (24.27) 4.12 (4.07) 6.03 (6.29)
(4) DDE249 12.11 (12.03) 3.07 (3.03) 4.67 (4.68)
(5) DDE250 29.34 (29.01) 4.42 (4.43) 6.21 (6.15)
(6) DDE173 16.02 (16.00) 3.21 (3.22) 7.57 (7.47)
HNMR analysis were performed on the synthesized compounds resulting in the values shown in table 2.
TABLE 2
Compound Solvent Chemical Shift (ppm)
(1) DDE246 D2O 2.97˜3.09(m, 4H, a), 3.86˜3.88(t, 4H, b)
(2) DDE247 D2O 2.82˜2.93(m, 4H, a), 3.85˜3.88(t, 4H, b)
(3) DDE248 D2O 1.57(s, 4H, c & d), 2.72˜2.79(m, 4H, a), 3.85˜
3.89(t, 4H, b)
(4) DDE249 D2O 1.90˜1.99(m, 4H, b), 2.92˜3.04(m, 4H, a),
3.69˜3.74(t, 4H, c)
(5) DDE250 DMSO-d6 0.96˜1.02(t, 2H, e′ & f), 1.11˜1.22(m, 6H, i &
j & d′ & g′), 1.44˜1.46(d, 2H, e & f), 1.78˜
1.82(d, 2H, d & g), 2.06(br, 2H, c & h),
5.22˜5.28(t, 2H, NaH′ & NbH′), 5.88˜5.90(d,
2H, NaH & NbH)
(6) DDE173 D2O 1.571 & 1.577(d, 4H, a & b)
EXAMPLE 3
In vitro cytotoxic activity of compounds (1)-(6) as synthesized and described in example 2 and of cisplatin was measured for human B-lineage acute lymphoblastic leukemia (NALM-6) and T-lineage acute lymphoblastic leukemia (MOLT-3) cells. The cells were incubated with concentrations ranging from 0.1 μM to 250 μM of the appropriate compound in 96-well plates for 48 hours. Cytotoxicity was determined by MTT [(3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide)] assays. The results are graphed in FIG. 1a for NALM-6 cells and FIG. 1b for MOLT-3 cells.
EXAMPLE 4
In vitro cytotoxic activity of compound (5) as synthesized and described in example 2 and of cisplatin was measured for human B-lineage acute lymphoblastic leukemia (NALM-6) cells. The cells were incubated with concentrations ranging from 0.2 μM to 20 μM of the appropriate compound in 96-well plates for 24 hours, 48 hours, 72 hours, and 96 hours. Cytotoxicity was determined by MTT assays and the IC50 values were determined using Graphpad Prism Software. The results are graphed in FIG. 2a for incubation of 24 hours, FIG. 2b for incubation of 48 hours, FIG. 2c for incubation of 72 hours, and FIG. 2d for incubation of 96 hours.
EXAMPLE 5
Various cells were incubated with the compounds (1)-(6) as synthesized and described in example 2 and with cisplatin in 96-well plates for 48 hours. Cytotoxicity was determined by MTT assays and the IC50 values were determined using Graphpad Prism Software. The results are shown in table 3.
TABLE 3
IC50 [MTT Assays] (μM)
MDA-
U266BL HS-Sultan BT-20 MB-231 PC3 U373
NALM-6 MOLT-3 (Multiple (Multiple (Breast (Breast (Prostate (Brain
Compound (Leukemia) (Leukemia) Myeloma) Myeloma) Cancer) Cancer) Cancer) Tumor)
(1) DDE246 >250 >250 >250 >250 >250 >250 >250 >250
(2) DDE247 60.1 ± 3.8 30.4 ± 4.3 35.6 ± 6.1 22.4 ± 2.8 51.4 ± 6.1 80.1 ± 5.5 91.3 ± 57.1  48.3 ± 2.8
(3) DDE248 75.3 ± 5.2 33.5 ± 4.5 N.D. N.D. 48.9 ± 7.3 N.D. 248.1 ± 9.3  129.5 ± 8.2
(4) DDE249 16.5 ± 6.5  9.5 ± 3.4 N.D. N.D. 81.3 ± 5.9 N.D. >250 >250
(5) DDE250  0.45 ± 0.02  0.66 ± 0.01 18.6 ± 2.1 21.5 ± 3.5 21.3 ± 3.1 38.5 ± 6.4 >250 >250
(6) DDE173 90.8 ± 6.2 41.8 ± 3.5 N.D. N.D. >250 N.D. >250 >250
Cisplatin  1.1 ± 0.02  1.04 ± 0.02 15.6 ± 3.5 31.8 ± 7.2 7.21 ± 3.5 51.6 ± 6.8 >250 >250
EXAMPLE 6
The effectiveness of inducing apoptosis by compound (5) as synthesized and described in example 2 was measured for human B-lineage acute lymphoblastic leukemia (NALM-6) cells, T-lineage acute lymphoblastic leukemia (MOLT-3) cells, and multiple myeloma (ARH-77) cells. The cells were incubated with concentrations of 5 μM of compound (5) in 96-well plates 48 hours. An in situ apoptosis assay, which allows the detection of exposed 3′-hydroxyl groups in fragmented DNA by TdT-mediated dUTP nick-end labeling (TUNEL), was used by fixing, permeabilizing and visualizing the DNA degradation. Apoptotic nuclei were detected visually with a color change. The amount of apoptotic nuclei of the control cells (not incubated with compound (5)) and the incubated cells were compared. In all cell lines, the compound (5) greatly increased apoptosis over the control.
EXAMPLE 7
The effect on clonogenic growth of human cancer cell lines was determined for compound (5) as synthesized and described in example 2 and cisplastin for human B-lineage acute lymphoblastic leukemia (NALM-6) cells and T-lineage acute lymphoblastic leukemia (MOLT-3) cells. The cells were incubated with concentrations ranging from 0 (control) to 10 μM of the appropriate compound for 24 hours at 37° C. in a humidified 5% CO2 atmosphere. The cells were then resuspended in clonogenic a-MEM medium containing 0.9% methyl cellulose. 1×104 cells/dish were plated in 35 mm Petri dishes and cultured for 7 days at 37° C. in a humidified 5% CO2 atmosphere. The colonies containing ≧20 cells were counted using an inverted phase microscope. The results are shown below in table 4.
TABLE 4
Concentra- Mean No. of %
Compound Cell Line tion (μM) Colonies/105 cells Inhibition
(5) DDE250 NALM-6  0 (control) 3696 (3056, 4336)
(5) DDE250 NALM-6  0.01 2110 (1928, 2292) 42.9
(5) DDE250 NALM-6  0.1 1908 (1886, 1930) 48.4
(5) DDE250 NALM-6  1  248 (228, 268) 93.3
(5) DDE250 NALM-6 10   0 >99.9
Cisplatin NALM-6  0 (control) 3696 (3056, 4336)
Cisplatin NALM-6  0.01 2044 (1996, 2092) 44.7
Cisplatin NALM-6  0.1 1832 (1744, 1920) 50.4
Cisplatin NALM-6  1  514 (470, 558) 86.0
Cisplatin NALM-6 10   0 >99.9
(5) DDE250 MOLT-3  0 (control) 3656 (3364, 3948)
(5) DDE250 MOLT-3  0.01 1182 (1080, 1284) 67.7
(5) DDE250 MOLT-3  0.1 1188 (1095, 1281) 67.5
(5) DDE250 MOLT-3  1  179 (170, 188) 95.1
(5) DDE250 MOLT-3 10   0 >99.9
Cisplatin MOLT-3  0 (control) 3656 (3364, 3948)
Cisplatin MOLT-3  0.01 1280 (1220, 1340) 64.9
Cisplatin MOLT-3  0.1 1168 (1116, 1220) 68.0
Cisplatin MOLT-3  1  241 (236, 246) 93.4
Cisplatin MOLT-3 10   0 >99.9
Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims (10)

What is claimed is:
1. A compound of formula I:
Figure US06737537-20040518-C00022
wherein:
R1 and R2 are independently chloro, iodo, or bromo,
R3, R4, R5 and R6 are independently hydrogen, hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, (C1-C6) alkoxy, (C1-C6) hydroxyalkyl, or aryl, wherein the aryl can be substituted with a halo, hydroxy, or (C1-C6) alkyl, and
a) when R1=R2=chloro
R7 and R8 are independently hydroxy, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, (C1-C6) alkoxy, methanol, n-propanol, isobutanol, t-butanol, octanol, decanol, tetradecanol, hexadecanol, aryl, wherein the aryl can be substituted with a halo, hydroxy, or (C1-C6) alkyl, or when taken together R7 and R8 form a group of the formula:
Figure US06737537-20040518-C00023
wherein m is 1 to 5 and R10 and R11 are independently hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, or (C1-C6) hydroxyalkyl, with the proviso that R10 and R11 are not both hydrogen or hydroxy when m=4; or
b) when R1=R2=iodo
R7 and R8 are independently hydroxy, (C2-C6) alkyl, (C3-C5) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, (C1-C6) alkoxy, (C5-C6) hydroxyalkyl, aryl, wherein the aryl can be substituted with a halo, hydroxy, or (C1-C6) alkyl, or when taken together R7 and R8 form a group of the formula:
Figure US06737537-20040518-C00024
wherein m is 1 to 5 and R10 and R11 are independently hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C2-C6) alkoxy, or (C1-C6) hydroxyalkyl, with the proviso that R10 and R11 are not both hydroxy when m=4; or
c) when R1=R2=bromo
R7 and R8 are independently hydroxy, (C3-C6) alkyl, (C3-C5) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, (C1-C6) alkoxy, (C2-C6) hydroxyalkyl, aryl, wherein the aryl can be substituted with a halo, hydroxy, or (C1-C6) alkyl, or when taken together R7 and R8 form a group of the formula:
Figure US06737537-20040518-C00025
wherein m is 1 to 5 and R10 and R11 are independently hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C2-C6) alkoxy, or (C2-C6) hydroxyalkyl, with the proviso that R10 and R11 are not both hydroxy when m=4; or
d) when R1≠R2,
R7 and R8 are independently hydroxy, (C1-C6) alkyl, (C3-C6) cycloalkyl, (C2-C6) alkenyl, (C3-C6) cycloalkenyl, (C1-C6) alkoxy, (C1-C6) hydroxyalkyl, aryl, wherein the aryl can he substituted with a halo, hydroxy, or (C1-C6) alkyl, or when taken together R7 and R8 form a group of the formula:
Figure US06737537-20040518-C00026
wherein m is 1 to 5 and R10 and R11 are independently hydrogen, halo, hydroxy, (C1-C6) alkyl, (C2-C6) alkenyl, (C1-C6) alkoxy, or (C1-C6) hydroxyalkyl, with the proviso that R10 and R11 are not both hydroxy when m=4;
or a pharmaceutically acceptable salt or ester thereof.
2. The compound of claim 1, wherein the R3, R4, R5, and R6 are independently hydrogen, hydroxy, or (C1-C3) alkyl.
3. The compound of claim 1, wherein R3, R4, R5, and R6 are hydrogen.
4. A compound:
Figure US06737537-20040518-C00027
or a pharmaceutically acceptable salt or ester thereof.
5. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.
6. A pharmaceutical composition comprising a compound of claim 4 and a pharmaceutically acceptable carrier.
7. A method of inducing apoptosis in a population of cells, comprising contacting a population of lymphoblastic leukemia cells, multiple myeloma cells, breast cancer cells, prostate cancer cells, or brain tumor cells with a compound according to claim 1.
8. The method of claim 7, wherein the compound is:
Figure US06737537-20040518-C00028
or a pharmaceutically acceptable salt or ester thereof.
9. The method of claim 15, wherein the compound is:
Figure US06737537-20040518-C00029
10. A method of inducing apoptosis in a population of cells, comprising contacting a population of lymphoblastic leukemia cells, multiple myeloma cells, breast cancer cells, prostate cancer cells, or brain tumor cells with a compound:
Figure US06737537-20040518-C00030
US10/146,971 1999-11-15 2002-05-15 Platinum (II) antitumor compounds Expired - Fee Related US6737537B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/146,971 US6737537B2 (en) 1999-11-15 2002-05-15 Platinum (II) antitumor compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16565299P 1999-11-15 1999-11-15
PCT/US2000/031297 WO2001036431A1 (en) 1999-11-15 2000-11-15 Diamino platinum (ii) antitumor complexes
US10/146,971 US6737537B2 (en) 1999-11-15 2002-05-15 Platinum (II) antitumor compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/031297 Continuation WO2001036431A1 (en) 1999-11-15 2000-11-15 Diamino platinum (ii) antitumor complexes

Publications (2)

Publication Number Publication Date
US20030176410A1 US20030176410A1 (en) 2003-09-18
US6737537B2 true US6737537B2 (en) 2004-05-18

Family

ID=22599852

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/146,971 Expired - Fee Related US6737537B2 (en) 1999-11-15 2002-05-15 Platinum (II) antitumor compounds

Country Status (3)

Country Link
US (1) US6737537B2 (en)
AU (1) AU1608801A (en)
WO (1) WO2001036431A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10141528B4 (en) * 2001-08-24 2006-08-10 Faustus Forschungs Cie. Translational Cancer Research Gmbh Platinum (II) and platinum (IV) complexes and their use
DE60229779D1 (en) * 2001-12-31 2008-12-18 Hans Rudolf Pfaendler LONG-TERM FAT ALCOHOL SUBSIDIES IN AGENTS AGAINST CANCER
DE102007060918A1 (en) 2007-12-14 2009-06-18 Rheinisch-Westfälische Technische Hochschule Aachen Process for the preparation of cycloplatinated platinum complexes, platinum complexes prepared by this process and their use
DK3049099T3 (en) 2013-09-26 2020-09-21 Priavoid Gmbh AMYLOID BETA-BINDING PEPTIDES AND ITS USE FOR THERAPY AND DIAGNOSIS OF ALZHEIMER'S DEMENTIA

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3892790A (en) 1972-04-10 1975-07-01 Rustenburg Platinum Mines Ltd Compositions containing platinum
US3904663A (en) 1972-04-10 1975-09-09 Rustenburg Platinum Mines Ltd Compositions containing platinum
US4115418A (en) 1976-09-02 1978-09-19 Government Of The United States Of America 1,2-diaminocyclohexane platinum (ii) complexes having antineoplastic activity
US4140707A (en) 1972-06-08 1979-02-20 Research Corporation Malonato platinum anti-tumor compounds
US4175133A (en) 1977-02-18 1979-11-20 United States Of America 1,2-Diaminocyclohexane platinum (II) complexes having antineoplastic activity against L1210 leukemia
US4228090A (en) 1978-04-20 1980-10-14 Johnson, Matthey & Co., Limited Compositions containing platinum
EP0130482A1 (en) 1983-06-20 1985-01-09 Research Corporation Diaminocyclohexane platinum complexes, process for preparing same and pharmaceutical compositions containing same
US4500465A (en) 1982-06-28 1985-02-19 Engelhard Corporation Solubilized platinum (II) complexes
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4587331A (en) 1984-12-17 1986-05-06 American Cyanamid Company Platinum complexes of polyhydroxylated alkylamines and 2-polyhydroxylated alkyl-1,2-diaminoethanes
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4661516A (en) 1983-06-20 1987-04-28 Research Corporation Diaminocyclohexane platinum complexes
US4670458A (en) 1986-01-31 1987-06-02 American Cyanamid Company Hydroxylated 1,2-diaminocyclohexane platinum complexes
US4758588A (en) 1983-06-20 1988-07-19 Research Corporation Technologies Diaminocyclohexane platinum complexes
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
EP0320960A1 (en) 1987-12-18 1989-06-21 Boehringer Mannheim Italia S.P.A. Amino anthracenediones-bis platinum complexes, useful as antitumoral agents
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US5011959A (en) 1986-11-17 1991-04-30 The Board Of Regents, The University Of Texas System 1,2-diaminocyclohexane-platinum complexes with antitumor activity
DE4041353A1 (en) 1990-12-21 1992-06-25 Bernhard K Dr Dr Keppler New complexes of platinum and crown ether carboxylic acid cpds. - with higher antitumour activity than cis-platin, lower toxicity and higher solubility in water
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3904663A (en) 1972-04-10 1975-09-09 Rustenburg Platinum Mines Ltd Compositions containing platinum
US3892790A (en) 1972-04-10 1975-07-01 Rustenburg Platinum Mines Ltd Compositions containing platinum
US4140707A (en) 1972-06-08 1979-02-20 Research Corporation Malonato platinum anti-tumor compounds
US4140707B1 (en) 1972-06-08 1989-12-19
US4115418A (en) 1976-09-02 1978-09-19 Government Of The United States Of America 1,2-diaminocyclohexane platinum (ii) complexes having antineoplastic activity
US4175133A (en) 1977-02-18 1979-11-20 United States Of America 1,2-Diaminocyclohexane platinum (II) complexes having antineoplastic activity against L1210 leukemia
US4228090A (en) 1978-04-20 1980-10-14 Johnson, Matthey & Co., Limited Compositions containing platinum
US4500465A (en) 1982-06-28 1985-02-19 Engelhard Corporation Solubilized platinum (II) complexes
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4758588A (en) 1983-06-20 1988-07-19 Research Corporation Technologies Diaminocyclohexane platinum complexes
EP0130482A1 (en) 1983-06-20 1985-01-09 Research Corporation Diaminocyclohexane platinum complexes, process for preparing same and pharmaceutical compositions containing same
US4661516A (en) 1983-06-20 1987-04-28 Research Corporation Diaminocyclohexane platinum complexes
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4587331A (en) 1984-12-17 1986-05-06 American Cyanamid Company Platinum complexes of polyhydroxylated alkylamines and 2-polyhydroxylated alkyl-1,2-diaminoethanes
US4670458A (en) 1986-01-31 1987-06-02 American Cyanamid Company Hydroxylated 1,2-diaminocyclohexane platinum complexes
US5011959A (en) 1986-11-17 1991-04-30 The Board Of Regents, The University Of Texas System 1,2-diaminocyclohexane-platinum complexes with antitumor activity
US5132323A (en) 1986-11-17 1992-07-21 Board Of Regents, The University Of Texas System 1,2-diaminocyclohexane-platinum complexes with antitumor activity
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
EP0320960A1 (en) 1987-12-18 1989-06-21 Boehringer Mannheim Italia S.P.A. Amino anthracenediones-bis platinum complexes, useful as antitumoral agents
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
DE4041353A1 (en) 1990-12-21 1992-06-25 Bernhard K Dr Dr Keppler New complexes of platinum and crown ether carboxylic acid cpds. - with higher antitumour activity than cis-platin, lower toxicity and higher solubility in water

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Cleare et al, Bioorganic Chem., 2, 187-210 (1973) Studies on the Antitumor Activity of Group VIII Transition Metal Complexes. Part I. Platinum (II).
Friedman et al., Chimica acta, 91 (1984) 75-80 Studies on the Inhibition of Fumarase and Malate Dehydrogenase by Second Generation Platinum Antitumor Drugs.
Zimmerman et al, Inorganica Chimica Acta, 292 (1999) 127-130 Synthesis and structures of (SP-4-2)-diiodobis(2-hydroxyethylamine)platinum(II), (SP-4-2)-dichlorobis (2-hydroxyethylamine)platinum(II) and (OC-622)-bis(2-hydroxyethylamine)tetrachloroplatinum(IV) in the crystal).

Also Published As

Publication number Publication date
WO2001036431A1 (en) 2001-05-25
AU1608801A (en) 2001-05-30
US20030176410A1 (en) 2003-09-18

Similar Documents

Publication Publication Date Title
US5736576A (en) Method of treating malignant tumors with thyroxine analogues having no significant hormonal activity
EP1177197B1 (en) Polyamines and their use in therapy
US20230382850A1 (en) Substituted bisphenylalkylurea compounds and methods of treating inflammatory conditions
FR2887882A1 (en) PYRIDO [2,3-D] PYRIMIDINE DERIVATIVES, THEIR PREPARATION, THEIR THERAPEUTIC APPLICATION
US9738613B2 (en) Substituted 1,2,3-triazoles as antitumor agents
US4882447A (en) Novel organic platinum complex and process for the preparation thereof
US6737537B2 (en) Platinum (II) antitumor compounds
US8252780B2 (en) Organometallic complexes as therapeutic agents
EP3129368B1 (en) Cytotoxic compounds which are inhibitors of the polymerisation of tubulin
CN108218800B (en) 1, 2, 3-triazole aminopeptidase N inhibitor and preparation method and application thereof
HU224839B1 (en) Substituted 1 betha-d-arabinofuranosylcytosine derivatives and use of the same for producing pharmaceutical compositions having antitumor activity
JP2007522165A (en) Compound having antitumor activity
EP1911451A1 (en) Protein-kinase CK2 inhibitors and their therapeutic applications
WO1989005637A1 (en) Improving toxicity profiles in chemotherapy
EP3947335A1 (en) Inhibitors of the n-terminal domain of the androgen receptor
US5728692A (en) Methotrexate derivative
US20040167180A1 (en) Methods for anti-tumor therapy
JP2009507858A (en) Quinoline derivatives and use as antitumor agents
FI75809B (en) FOERFARANDE FOER FRAMSTAELLNING AV THERAPEUTIC ACTIVATED VACCINE DERIVATIVES AV 6,6'-METHYLENE-BIS (2,2,4-TRIMETYL-1,2-DIHYDROQUINOLINE).
US4423076A (en) 1-Branched-alkyl-3-(2-haloethyl)-3-nitrosoureas as novel antitumor agents
KR100249408B1 (en) (s)-2,3--dihydropolyprenol, (s)-2,3-dihydropolyprenyl monophosphate, of preparing the same and agents for inhibiting the growth or metascancers comprising the same
JPS62167744A (en) Fluorinated diaminoalkine derivative
US4820706A (en) Pteridine derivatives and method of treating leukemia employing same
US4920126A (en) Barbituric acid derivative and treatment of leukemia and tumors therewith
EP3967683A1 (en) Substituted fluorine-containing imidazole salt compound, preparation method therefor, pharmaceutical composition thereof and use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: KABUSHIKI KAISHA HAYASHIBARA SEIBUTSU KAGAKU KENKY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KINUGASA, KAZUSHI;MANDAI, SHINYA;REEL/FRAME:013285/0120

Effective date: 20020401

AS Assignment

Owner name: PARKER HUGHES INSTITUTE, MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:UCKUN, FATIH M.;NARLA, RAMA K.;REEL/FRAME:013448/0254;SIGNING DATES FROM 20020924 TO 20021023

CC Certificate of correction
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20080518